Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 324(4): C927-C940, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36717099

ABSTRACT

Ferroptosis is the ideal therapeutic target for hepatic ischemia and reperfusion injury (HIRI). The µ opioid receptor (MOR) is associated with ferroptosis in HIRI. We aimed to determine the ferroptosis-related therapeutic mechanism of MOR in HIRI. A model of HIRI was established in BALB/c mice. Primary hepatocytes isolated from mice were stimulated by hypoxia/reoxygenation (H/R). Changes in histopathology were determined by H&E staining. Alterations in ferroptosis were evaluated by malondialdehyde (MDA), iron, glutathione (GSH), ACSL4, GPX4, and mitochondrial morphology. ALT and AST were used to determine hepatic function. First, we found that hepatic ischemia/reperfusion (I/R) induced the destruction of hepatic tissue structure and dead hepatocytes and determined that ferroptosis occurred in vivo and in vitro. During HIRI, the expression levels of HIF-1α and KCNQ1OT1 were significantly upregulated. We demonstrated that sufentanil improved the damage in the liver and hepatocytes undergoing I/R. Importantly, sufentanil inhibited ferroptosis in HIRI. In addition, sufentanil downregulated the expression levels of HIF-1α and KCNQ1OT1 in HIRI. Increases in HIF-1α and KCNQ1OT1 reversed the role of sufentanil in ferroptosis and HIRI. Subsequently, we determined that HIF-1α could activate the transcription of KCNQ1OT1 by binding to its promoter. In addition, KCNQ1OT1 was demonstrated to enhance ACSL4 stability by interacting with SRSF1. Finally, we observed that KCNQ1OT1 downregulation protected hepatocytes from hepatic I/R and inhibited ferroptosis. KCNQ1OT1 upregulation aggravated ferroptosis and hepatic injury during I/R. However, decreases in ACSL4 and SRSF1 reversed the harmful role of KCNQ1OT1 upregulation in HIRI. MOR alleviated ferroptosis in HIRI via the HIF-1α/KCNQ1OT1 axis.


Subject(s)
Ferroptosis , Hypoxia-Inducible Factor 1, alpha Subunit , Receptors, Opioid, mu , Reperfusion Injury , Animals , Mice , Ferroptosis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/metabolism , Liver/metabolism , Liver/pathology , Receptors, Opioid, mu/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Sufentanil/metabolism , Sufentanil/therapeutic use , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
2.
Comput Math Methods Med ; 2022: 6267720, 2022.
Article in English | MEDLINE | ID: mdl-35356663

ABSTRACT

Objective: Sufentanil is the most common drug in clinical practice for the treatment of ischemic heart disease. This study is to investigate the protective mechanism of sufentanil on rat myocardial ischemia-reperfusion (I/R) injury. Methods: A rat I/R model was established by ligating the left anterior descending coronary artery. A total of 24 SD male rats were enrolled and divided randomly into the control group, I/R group, sufentanil group (SUF; 3 µg/kg), and diltiazem group (DLZ; 20 mg/kg; positive control). The rat hearts were subjected to 30 min of ischemia followed by 120 min of reperfusion. Subsequently, hemodynamics, pathological changes of myocardial tissue, serum biochemical parameters, oxidative stress factors, the level of serum inducible nitric oxide synthases (iNOS), interleukin-6 (IL-6), and other bioactive factors were analyzed in the rats. Result: Compared with the I/R group, sufentanil significantly improved cardiac action, myocardial fiber, and cardiomyocyte morphology and reduced inflammatory cell infiltration in rats in the SUF group. And the level of creatine kinase isoenzyme (CK-MB), troponin (cTn), lactate dehydrogenase (LDH), malondialdehyde (MDA), iNOS, and IL-6 was significantly declined in the serum of SUF group, while the activities of glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) were significantly activated in the myocardial tissues. In addition, sufentanil also significantly decreased the protein expression of GRP78, CHOP, Caspase 12, and ATF6 in the myocardial tissue of the SUF group. Conclusion: Sufentanil has a significant protective activity on myocardial I/R injury in rats, the mechanism of which may be associated with the inhibition of endoplasmic reticulum stress and oxidative stress.


Subject(s)
Myocardial Reperfusion Injury , Animals , Endoplasmic Reticulum Stress , Humans , Male , Malondialdehyde , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Rats , Sufentanil/metabolism , Sufentanil/pharmacology , Sufentanil/therapeutic use
3.
Eur J Histochem ; 66(1)2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35016494

ABSTRACT

Stroke is a brain system disease with a high fatality rate and disability rate. About 80% of strokes are ischemic strokes. Cerebral ischemia-reperfusion injury (CIRI) caused by ischemic stroke seriously affects the prognosis of stroke patients. The purpose of this study is to investigate the effect of sufentanil (SUF) on CIRI model rats. We used middle cerebral artery occlusion (MCAO) to make the CIRI model in rats and monitored region cerebral blood flow (rCBF) to ensure that blood flow was blocked and recanalized. We used ELISA and RT-PCR to detect the expression of inflammatory factors in rat serum and brain tissue. In addition, we detected the expression of metalloproteinase (MMP) 2, MMP9 and collagen IV in brain tissues and performed Evans blue (EB) assay to determine the permeability of the blood-brain barrier (BBB). Finally, we clarified the apoptosis of brain tissue through the TUNEL staining and the detection of caspase3, Bcl2 and Bax. Various concentrations of SUF, especially 5, 10 and 25 µg/kg of SUF, all alleviated the infarct size, neurological function and brain edema of MCAO rats. SUF pretreatment also effectively reduced the expression of inflammatory cytokines in MCAO rats, including interleukin (IL)-1ß, IL-4, IL-6, IL-8, IL-10 and tumor necrosis factor (TNF)-α. In addition, SUF also inhibited MMP2 and MMP9 and promoted the expression of collagen IV, indicating that SUF attenuated the destruction of the BBB. SUF also inhibited caspase3 and Bax rats and promoted Bcl2 in MCAO rats, thus inhibiting cell apoptosis. SUF pretreatment effectively improved the neurological function and cerebral infarction of MCAO rats, inhibited excessive inflammation in rats, protected the BBB, and inhibited cell apoptosis in brain tissue.


Subject(s)
Brain Ischemia , Reperfusion Injury , Animals , Blood-Brain Barrier/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Humans , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Inflammation/drug therapy , Rats , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Sufentanil/metabolism , Sufentanil/pharmacology
4.
Drugs R D ; 20(1): 1-10, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31820365

ABSTRACT

BACKGROUND: The application of modeling and simulation approaches in clinical pharmacology studies has gained momentum over the last 20 years. OBJECTIVES: The objective of this study was to develop six empirical models from clearance data obtained from children aged > 2 years and adults to evaluate the suitability of the models to predict drug clearance in children aged ≤ 2 years (preterm, term, and infants). METHODS: Ten drugs were included in this study and administered intravenously: alfentanil, amikacin, busulfan, cefetamet, meperidine, oxycodone, propofol, sufentanil, theophylline, and tobramycin. These drugs were selected according to the availability of individual subjects' weight, age, and clearance data (concentration-time data for these drugs were not available to the author). The chosen drugs are eliminated by extensive metabolism by either the renal route or both the renal and hepatic routes. The six empirical models were (1) age and body weight-dependent sigmoidal maximum possible effect (Emax) maturation model, (2) body weight-dependent sigmoidal Emax model, (3) uridine 5'-diphospho [body weight-dependent allometric exponent model (BDE)], (4) age-dependent allometric exponent model (ADE), (5) a semi-physiological model, and (6) an allometric model developed from children aged > 2 years to adults. The model-predicted clearance values were compared with observed clearance values in an individual child. In this analysis, a prediction error of ≤ 50% for mean or individual clearance values was considered acceptable. RESULTS: Across all age groups and the ten drugs, data for 282 children were compared between observed and model-predicted clearance values. The validation data consisted of 33 observations (sum of different age groups for ten drugs). Only three of the six models (body weight-dependent sigmoidal Emax model, ADE, and semi-physiological model) provided reasonably accurate predictions of clearance (> 80% observation with ≤ 50% prediction error) in children aged ≤ 2 years. In most instances, individual predicted clearance values were erratic (as indicated by % error) and were not in agreement with the observed clearance values. CONCLUSIONS: The study indicated that simple empirical models can provide more accurate results than complex empirical models.


Subject(s)
Metabolic Clearance Rate , Models, Biological , Adult , Alfentanil/administration & dosage , Alfentanil/metabolism , Amikacin/administration & dosage , Amikacin/metabolism , Busulfan/administration & dosage , Busulfan/metabolism , Ceftizoxime/administration & dosage , Ceftizoxime/analogs & derivatives , Ceftizoxime/metabolism , Child, Preschool , Humans , Infant , Injections, Intravenous , Meperidine/administration & dosage , Meperidine/metabolism , Oxycodone/administration & dosage , Oxycodone/metabolism , Propofol/administration & dosage , Propofol/metabolism , Sufentanil/administration & dosage , Sufentanil/metabolism , Theophylline/administration & dosage , Theophylline/metabolism , Tobramycin/administration & dosage , Tobramycin/metabolism
5.
Chem Biol Drug Des ; 91(4): 902-914, 2018 04.
Article in English | MEDLINE | ID: mdl-29222847

ABSTRACT

Novel substituted amino acid tethered norsufentanil derivatives were synthesized by the four-component Ugi reaction. Norsufentanil was reacted with succinic anhydride to produce the corresponding carboxylic acid. The resulting carboxylic acid has undergone a multicomponent reaction with different aldehydes, amines, and isocyanides to produce a library of the desired compounds. In all cases, amide bond rotation was observed in the NMR spectra. In vivo analgesic activity of the synthesized compounds was evaluated by a tail flick test. Very encouraging results were obtained for a number of the synthesized products. Some of the synthesized compounds such as 5a, 5b, 5h, 5j, and 5r were found to be more potent than sufentanil, sufentanil citrate, and norsufentanil. Binding modes between the compounds and mu and delta-opioid receptors were studied by molecular docking method. The relationship between the molecular structural features and the analgesic activity was investigated by a quantitative structure-activity relationship model. The results of the molecular modeling studies and the in vivo analgesic activity suggested that the majority of the synthesized compounds were more potent than sufentanil and norsufentanil.


Subject(s)
Analgesics/chemical synthesis , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Sufentanil/analogs & derivatives , Acute Pain/drug therapy , Analgesics/metabolism , Analgesics/therapeutic use , Animals , Binding Sites , Male , Mice , Naloxone/chemistry , Naloxone/metabolism , Protein Structure, Tertiary , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Sufentanil/chemistry , Sufentanil/metabolism , Sufentanil/therapeutic use
6.
Clin Pharmacol Ther ; 104(1): 188-200, 2018 07.
Article in English | MEDLINE | ID: mdl-29027194

ABSTRACT

The accuracy of physiologically based pharmacokinetic (PBPK) model prediction in children, especially those younger than 2 years old, has not been systematically evaluated. The aim of this study was to characterize the pediatric predictive performance of the PBPK approach for 10 drugs extensively metabolized by CYP1A2 (theophylline), CYP2C8 (desloratidine, montelukast), CYP2C9 (diclofenac), CYP2C19 (esomeprazole, lansoprazole), CYP2D6 (tramadol), and CYP3A4 (itraconazole, ondansetron, sufentanil). Model performance in children was evaluated by comparing simulated plasma concentration-time profiles with observed clinical results for each drug and age group. PBPK models reasonably predicted the pharmacokinetics of desloratadine, diclofenac, itraconazole, lansoprazole, montelukast, ondansetron, sufentanil, theophylline, and tramadol across all age groups. Collectively, 58 out of 67 predictions were within 2-fold and 43 out of 67 predictions within 1.5-fold of observed values. Developed PBPK models can reasonably predict exposure in children age 1 month and older for an array of predominantly CYP metabolized drugs.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Acetates/metabolism , Acetates/pharmacokinetics , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacokinetics , Anti-Asthmatic Agents/metabolism , Anti-Asthmatic Agents/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antifungal Agents/metabolism , Antifungal Agents/pharmacokinetics , Bronchodilator Agents/metabolism , Bronchodilator Agents/pharmacokinetics , Child , Child, Preschool , Cyclopropanes , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/metabolism , Diclofenac/metabolism , Diclofenac/pharmacokinetics , Esomeprazole/metabolism , Esomeprazole/pharmacokinetics , Histamine H1 Antagonists, Non-Sedating/metabolism , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Humans , Infant , Infant, Newborn , Itraconazole/metabolism , Itraconazole/pharmacokinetics , Lansoprazole/metabolism , Lansoprazole/pharmacokinetics , Loratadine/analogs & derivatives , Loratadine/metabolism , Loratadine/pharmacokinetics , Ondansetron/metabolism , Ondansetron/pharmacokinetics , Proton Pump Inhibitors/metabolism , Proton Pump Inhibitors/pharmacokinetics , Quinolines/metabolism , Quinolines/pharmacokinetics , Serotonin Antagonists/metabolism , Serotonin Antagonists/pharmacokinetics , Sufentanil/metabolism , Sufentanil/pharmacokinetics , Sulfides , Theophylline/metabolism , Theophylline/pharmacokinetics
7.
Br J Anaesth ; 113(4): 677-87, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25001621

ABSTRACT

BACKGROUND: Our objective was to evaluate the effect of intensive care treatment on the protein binding of sufentanil and hydromorphone in cardiac surgery patients during postoperative analgesia using a target-controlled infusion (TCI) and patient-controlled analgesia (PCA). METHODS: Fifty adult patients were enrolled in this prospective randomized study; of which, 49 completed the study (age range 40-81 yr). Sufentanil was administered as an analgesic intraoperatively, and hydromorphone was dosed after operation with TCI and PCA until 8 a.m. on the first postoperative day. Arterial plasma samples were collected for drug and protein concentration measurements up to 24 h after cardiac surgery. Corresponding patient data were collected from the electronic patient data system. After explorative data analysis with principal component analysis, multivariate regression analysis and non-linear mixed effects modelling was used to study the effect of treatment on protein binding. RESULTS: Data of 35 patients were analysed. The median protein binding of sufentanil and hydromorphone was 88.4% (IQ range 85.7-90.5%) and 11.6% (IQ range 9.5-14.3%), respectively. Free fraction of sufentanil increased towards the end of the study period, whereas hydromorphone free fraction remained nearly constant. The total sufentanil concentration and volume balance were identified as significant covariates for the protein binding of sufentanil. For the protein binding of hydromorphone, no significant covariate effects were found. CONCLUSIONS: Sufentanil protein binding was significantly dependent on changes in the total drug concentration and volume balance addressing the importance of adequate dosing and fluid-guided therapy. Hydromorphone protein binding was nearly constant throughout the study period. CLINICAL TRIAL REGISTRATION: EudraCT 2011-003648-31 and ClinicalTrials.gov: NCT01490268.


Subject(s)
Analgesics, Opioid/metabolism , Analgesics, Opioid/therapeutic use , Cardiac Surgical Procedures/methods , Critical Care , Hydromorphone/metabolism , Hydromorphone/therapeutic use , Pain, Postoperative/drug therapy , Sufentanil/metabolism , Sufentanil/therapeutic use , Adult , Aged , Algorithms , Analgesia, Patient-Controlled , Analgesics, Opioid/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Hydromorphone/administration & dosage , Male , Middle Aged , Nonlinear Dynamics , Prospective Studies , Protein Binding , Regression Analysis , Sufentanil/administration & dosage , Thoracotomy
9.
Pain ; 126(1-3): 234-44, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16949748

ABSTRACT

Lidocaine-like sodium channel blocking drugs provide pain relief either by interrupting impulse conduction in neurons when applied locally in high concentrations or, when given systemically, by suppressing high-frequency ectopic discharges due to preferential drug binding to inactivated channel states. Lidocaine-like actions of opioids have frequently been demonstrated clinically. However, drug binding to resting and inactivated channel conformations has been studied systematically only in the case of meperidine. The aim of this in vitro study was to investigate the effects of four currently used opioids on heterologously expressed neuronal (NaV(1.2)) voltage-gated sodium channels. Block of sodium currents was studied at hyperpolarized holding potentials and at depolarized potentials inducing either fast- or slow-inactivation. Sufentanil, fentanyl and tramadol but not morphine reversibly suppressed sodium inward currents at high concentrations (half-maximum blocking concentrations (IC50) 49+/-4, 141+/-6 and 103+/-8 microM) when depolarizations were started from hyperpolarized holding potentials. Short depolarizations inducing fast-inactivation and long prepulses inducing slow-inactivation significantly (*p < or = 0.001) increased the blocking potency for these opioids. 15% slow inactivated channels reduced the respective IC50 values to 5+/-3, 12+/-2 and 21+/-2 microM. These results show that: (1) Sufentanil, fentanyl and tramadol block voltage-gated sodium channels with half-maximum inhibitory concentrations similar to the IC50 reported for meperidine. (2) Slow inactivation--a physiological mechanism to suppress ectopic activity in response to slow shifts in membrane potential--increases binding affinity for sufentanil, fentanyl and tramadol. (3) Morphine has no such effects.


Subject(s)
Analgesics, Opioid/pharmacology , Fentanyl/pharmacology , Morphine/pharmacology , Nerve Tissue Proteins/drug effects , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology , Sufentanil/pharmacology , Tramadol/pharmacology , Analgesics, Opioid/metabolism , Cell Line , Electric Conductivity , Fentanyl/metabolism , Humans , Ion Channel Gating , NAV1.2 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , Sodium Channel Blockers/metabolism , Sodium Channels/drug effects , Sodium Channels/metabolism , Sufentanil/metabolism , Tramadol/metabolism
10.
Bioorg Med Chem Lett ; 15(7): 1773-7, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15780604

ABSTRACT

The synthesis of an (18)F-labeled sufentanil analogue with apparent high mu-opioid receptor selectivity is reported. Intravenous injection of N-[4-(methoxymethyl)-1-[2-(2-thienyl)ethyl]-4-piperidinyl]-N-phenyl-2-(+/-)-[(18)F]fluoropropan-amide in mice resulted in high brain uptake and a regional brain activity distribution corresponding to the mu-opioid receptor expression pattern. The developed ligand is a promising tracer for extended protocols in mu-opioid receptor mapping and quantitation with positron emission tomography.


Subject(s)
Fluorine Radioisotopes , Radiopharmaceuticals/metabolism , Receptors, Opioid, mu/metabolism , Sufentanil/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Mice , Positron-Emission Tomography/methods , Sufentanil/analogs & derivatives
11.
Anesthesiology ; 96(4): 913-20, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11964599

ABSTRACT

BACKGROUND: The efflux transporter P-glycoprotein, a member of the adenosine triphosphate-binding cassette superfamily, is a major determinant of the pharmacokinetics and pharmacodynamics of the opioid loperamide, a well-recognized antidiarrheal agent. Animal studies indicate that P-glycoprotein limits morphine entry into the brain. In this study, the authors examined whether other opioids of importance to anesthesiologists such as fentanyl, sufentanil, and alfentanil, and also morphine-6-glucuronide and morphine-3-glucuronide, are P-glycoprotein substrates and whether, in turn, these opioids act also as P-glycoprotein inhibitors. METHODS: The transcellular movement of the various opioids, including loperamide and morphine, was assessed in L-MDR1 (expressing P-glycoprotein) and LLC-PK1 cell monolayers (P-glycoprotein expression absent). A preferential basal-to-apical versus apical-to-basal transport in the L-MDR cells but not the LLC-PK1 cells is seen for P-glycoprotein substrates. In addition, the effect of the various opioids on the transcellular movement of the prototypical P-glycoprotein substrate digoxin was examined in Caco-2 cell monolayers. IC50 values were calculated according to the Hill equation. RESULTS: Loperamide was a substrate showing high dependence on P-glycoprotein in that basal-apical transport was nearly 10-fold greater than in the apical-basal direction in L-MDRI cells. Morphine also showed a basal-to-apical gradient in the L-MDR1 cell monolayer, indicating that it too is a P-glycoprotein substrate, but with less dependence than loperamide in that only 1.5-fold greater basal-apical directional transport was observed. Fentanyl, sufentanil, and alfentanil did not behave as P-glycoprotein substrates, whereas the morphine glucuronides did not cross the cell monolayers at all, whether P-glycoprotein was present or not. Loperamide, sufentanil, fentanyl, and alfentanil inhibited P-glycoprotein-mediated digoxin transport in Caco-2 cells with IC50 values of 2.5, 4.5, 6.5, and 112 microm, respectively. Morphine and its glucuronides (20 microm) did not inhibit digoxin (5 microm) transport in Caco-2 cells, and therefore IC50 values were not determined. CONCLUSIONS: Opioids have a wide spectrum of P-glycoprotein activity, acting as both substrates and inhibitors, which might contribute to their varying central nervous system-related effects.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Alfentanil/metabolism , Analgesics, Opioid/metabolism , Antidiarrheals/metabolism , Fentanyl/metabolism , Loperamide/metabolism , Morphine/metabolism , Sufentanil/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Alfentanil/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Fentanyl/pharmacology , Humans , Loperamide/pharmacology , Morphine/pharmacology , Sufentanil/pharmacology
12.
Anesth Analg ; 93(3): 543-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11524316

ABSTRACT

Because some clinical studies have suggested that opioids used in anesthesia may have different deleterious hemodynamic effects, we compared the direct myocardial effects of cumulative concentrations of remifentanil, sufentanil, fentanyl, and alfentanil on inotropic and lusitropic variables of isolated human myocardium in vitro. Human right atrial trabeculae, obtained from patients scheduled for coronary bypass surgery or aortic valve replacement, were suspended vertically in an oxygenated (95% oxygen/5% CO(2)) Tyrode's modified solution ([Ca(2+)](o) = 2.0 mM, 37 degrees C, pH 7.40, stimulation frequency 1 Hz). The effects of cumulative concentrations (10(-11), 10(-10), 10(-9), 10(-8), 10(-7), and 10(-6) M) of remifentanil (n = 8), sufentanil (n = 8), fentanyl (n = 8), and alfentanil (n = 8) on inotropic and lusitropic variables of isometric twitches were measured. Remifentanil, sufentanil, and fentanyl did not modify active isometric force and peak of the positive force derivative as compared with the Control group. Alfentanil induced a dose-dependent decrease in active isometric force and peak of the positive force derivative. This effect was abolished in the presence of [Ca(2+)](o) = 4.0 mM. None of these opioids altered lusitropic variables.


Subject(s)
Alfentanil/pharmacology , Anesthetics, Intravenous/pharmacology , Atrial Function, Right/drug effects , Fentanyl/pharmacology , Heart/drug effects , Piperidines/pharmacology , Sufentanil/pharmacology , Aged , Alfentanil/metabolism , Anesthetics, Intravenous/metabolism , Female , Fentanyl/metabolism , Heart Atria/drug effects , Heart Atria/metabolism , Humans , In Vitro Techniques , Male , Middle Aged , Muscle Relaxation/drug effects , Myocardial Contraction/drug effects , Myocardium/metabolism , Piperidines/metabolism , Remifentanil , Sufentanil/metabolism
13.
J Neurochem ; 73(1): 289-300, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10386982

ABSTRACT

The influence of membrane microviscosity on mu-opioid agonist and antagonist binding, as well as agonist efficacy, was examined in membranes prepared from SH-SY5Y cells and from a C6 glioma cell line stably expressing the rat mu-opioid receptor (C6mu). Addition of cholesteryl hemisuccinate (CHS) to cell membranes increased membrane microviscosity and reduced the inhibitory effect of sodium and guanine nucleotides on the affinity of the full agonists sufentanil and [D-Ala2,N-MePhe4,Gly-ol5]enkephalin (DAMGO) for the mu-opioid receptor. Binding of the antagonists [3H]naltrexone and [3H]diprenorphine and the partial agonist nalbuphine was unaffected by CHS. The effect of CHS on agonist binding was reversed by subsequent addition of cis-vaccenic acid, suggesting that the effect of CHS is the result of increased membrane microviscosity and not a specific sterol-receptor interaction. CHS addition increased the potency of DAMGO to stimulate guanosine-5'-O-(3-[35S]thio)triphosphate binding by fourfold, whereas the potency of nalbuphine was unaffected. However, nalbuphine efficacy relative to that of the full agonist DAMGO was strongly increased in CHS-treated membranes compared with that in control membranes. Membrane rigidification also resulted in an increased efficacy for the partial agonists meperidine, profadol, and butorphanol relative to that of DAMGO as measured by agonist-stimulated GTPase activity in control and CHS-modified membranes. These findings support a regulatory role for membrane microviscosity in receptor-mediated G protein activation.


Subject(s)
Cell Membrane/physiology , Membrane Fluidity/physiology , Narcotics/metabolism , Protein Conformation , Receptors, Opioid, mu/chemistry , Animals , Cell Line , Cell Membrane/drug effects , Cholesterol Esters/pharmacology , Diprenorphine/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/metabolism , Glioma , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Nalbuphine/metabolism , Naltrexone/metabolism , Narcotic Antagonists/metabolism , Neuroblastoma , Rats , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Sodium Chloride/pharmacology , Sufentanil/metabolism , Tumor Cells, Cultured , Viscosity
14.
Pharmacol Toxicol ; 85(6): 263-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10628901

ABSTRACT

Fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), significantly potentiates analgesia when administered in animals together with opioids. The aim of the present study was to investigate the effects of fluvoxamine on sufentanil antinociception and tolerance. Following animal care committee approval, the effects of continuous infusions of fluvoxamine and sufentanil were studied in behavioural tests (hot-plate test, tail-flick test, catalepsy test) in Sprague-Dawley rats with a jugular vein catheter. Saline was administered as a control. The time-effect curves for continuous intravenous sufentanil indicate dose-related antinociception and rapid development of tolerance in the hot-plate and tail-flick tests. Co-administration of fluvoxamine with continuous sufentanil enhances antinociception and attenuates development of tolerance, most clearly seen in the tail-flick test. Fluvoxamine alone and saline were not effective. No animal showed catalepsy. As a side effect we observed a marked loss of body weight. The IC50 values of sufentanil binding with and without fluvoxamine addition are 0.56+/-0.17 nM and 0.3+/-0.15 nM, respectively, indicating no direct effect on the occupancy of sufentanil on the mu-receptor by this serotonin reuptake inhibitor. In conclusion, we were able to show that the combination of an opioid with an SSRI at low doses improves analgesia and decreases development of tolerance in nociceptive tests in rats. The clinical implications of these promising results in an animal model, however, await further investigation.


Subject(s)
Analgesics, Opioid/pharmacology , Fluvoxamine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Sufentanil/pharmacology , Analgesics, Opioid/metabolism , Animals , Dose-Response Relationship, Drug , Drug Tolerance , Fluvoxamine/administration & dosage , Fluvoxamine/metabolism , Infusions, Intravenous , Male , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/metabolism , Sufentanil/metabolism
15.
Eur J Pharmacol ; 354(2-3): 227-37, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9754924

ABSTRACT

The apparent thermodynamic parameters of binding of ten ligands to the cloned rat mu-opioid receptor stably expressed in Chinese hamster ovary (CHO) cells were investigated. For every ligand, the Kd or Ki values at 0 degrees C, 12 degrees C, 25 degrees C and 37 degrees C were determined, a van't Hoff plot was generated and deltaH degrees' , deltaS degrees' and -TdeltaS degrees' and deltaG degrees' were calculated. Changes in free energy (deltaG degrees') ranged from -10.35 to -15.65 kcal/mol. The binding of sufentanil, ohmefentanyl, diprenorphine and D-Phe-Cys-Tyr-D-Trp-Arg-Thr-penicillamineThr-NH2 (CTAP) was endothermic (deltaH degrees' > 0) and driven by an increase in entropy (-TdeltaS degrees' = -13.08 to -18.57 kcal/mol). The binding of naltrexone was exothermic (deltaH degrees' = -12.56 kcal/mol) and essentially enthalpy-driven. The binding of morphine, methadone, pentazocine, [D-Ala2, NMePhe4, Gly-ol]enkephalin (DAMGO) and Tyr-Pro-NMePhe-D-Pro-NH2 (PL017) was exothermic (deltaH degrees' = -3.53 to -9.95 kcal/mol) and occurred with an increase in entropy (-TdeltaS degrees' = -2.48 to -7.92 kcal/mol). Plots of enthalpy versus entropy and enthalpy versus free energy were linear, although enthalpy-entropy compensation was not evident. The entropy changes were not correlated with apparent lipophilicity of the compounds. These results suggest that: (1) opioid ligands bind to the mu receptor by specific mechanisms, unrelated to lipid solubility; (2) the mechanism of binding is not universally different for peptide and non-peptide ligands; (3) the nature of binding does not a priori determine intrinsic activity. The results reveal a novel differentiation of opioid ligands into two groups (group 1: ohmefentanyl, sufentanil, diprenorphine, CTAP and PL017; group 2: naltrexone, morphine, methadone, DAMGO, pentazocine), based on two distinct relationships between enthalpy versus free energy of binding, the details of which are yet to be elucidated.


Subject(s)
Receptors, Opioid, mu/metabolism , Analgesics/pharmacology , Animals , Binding, Competitive , CHO Cells , Cricetinae , Diprenorphine/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/pharmacology , Fentanyl/analogs & derivatives , Fentanyl/metabolism , Ligands , Methadone/metabolism , Morphine/metabolism , Naltrexone/metabolism , Narcotic Antagonists/pharmacology , Pentazocine/metabolism , Peptide Fragments , Peptides/pharmacology , Radioligand Assay , Rats , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Somatostatin , Sufentanil/metabolism , Thermodynamics , Time Factors
16.
J Chromatogr Sci ; 35(10): 461-6, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9336954

ABSTRACT

A sensitive, specific urinary assay for fentanyl, sufentanil, and alfentanil based on their N-dealkylated metabolites is described. Norfentanyl, norsufentanil-noralfentanil, and 2H5-norfentanyl are synthesized and characterized by standard analytical techniques. Derivatization of these secondary amines to yield the pentafluorobenzamides produces stable products with good gas chromatographic properties and unique, high-mass fragments in their mass spectra. These properties are utilized to develop a drug screening procedure based on gas chromatography-mass spectrometry to detect these major metabolites in human urine. The metabolites are isolated from urine samples by a liquid-liquid extraction procedure. The method allows for detection of metabolite concentrations as low as 0.3 ng/mL.


Subject(s)
Alfentanil/urine , Analgesics, Opioid/urine , Drug Residues/analysis , Fentanyl/urine , Gas Chromatography-Mass Spectrometry/methods , Narcotics/urine , Sufentanil/urine , Alfentanil/chemistry , Alfentanil/metabolism , Analgesics, Opioid/chemistry , Analgesics, Opioid/metabolism , Fentanyl/analogs & derivatives , Fentanyl/chemistry , Fentanyl/metabolism , Humans , Narcotics/chemistry , Narcotics/metabolism , Sensitivity and Specificity , Sufentanil/analogs & derivatives , Sufentanil/chemistry , Sufentanil/metabolism
17.
Anesth Analg ; 84(6): 1262-8, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9174304

ABSTRACT

This study investigated factors that influence the placental transfer of sufentanil using the dual-perfused, single-cotyledon human placental model. Placentas were collected from healthy women. Experiments were designed to elucidate the effects of maternal protein binding, changing maternal sufentanil concentration (1, 10, 20, and 100 ng/mL) and decreasing fetal pH (fetal acidemia 7.2, 7.0, 6.8) on the placental transfer of sufentanil. Sufentanil crossed the placenta rapidly at a rate two-thirds that of the transfer marker, antipyrine. Sufentanil transfer increased linearly with the maternal concentration (r = 0.999). Sufentanil/antipyrine maternal to fetal (M-->F) transfer ratios were significantly reduced (0.66 +/- 0.05 vs 0.40 +/- 0.04, P < 0.05) when fresh frozen plasma was added to the maternal circuit to enhance protein binding. Fetal pH and sufentanil transfer were related because sufentanil M-->F clearance increased significantly as the fetal pH decreased (r = 0.973, P < 0.05). Sufentanil appears to cross the placenta by passive diffusion but is modulated by the degree of maternal protein binding. Sufentanil M-->F transfer is enhanced by fetal acidemia.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Fetus/metabolism , Maternal-Fetal Exchange , Placenta/metabolism , Sufentanil/pharmacokinetics , Adult , Analgesics, Opioid/metabolism , Blood Proteins/metabolism , Female , Humans , Models, Biological , Pregnancy , Protein Binding , Sufentanil/metabolism
18.
Biochem Pharmacol ; 53(11): 1613-9, 1997 Jun 01.
Article in English | MEDLINE | ID: mdl-9264313

ABSTRACT

Fentanyl, sufentanil, and alfentanil are commonly used as opioid analgesics. Alfentanil clearance has previously been shown to exhibit an important interindividual variability, which was not observed for fentanyl or sufentanil. Differences in pharmacokinetic parameters of alfentanil have previously been associated with the wide distribution of CYP3A4, the only known hepatic cytochrome P450 monooxygenase (CYP) involved in the conversion of alfentanil to noralfentanil. Little is known about the involvement of CYP enzymes in the oxidative metabolism of fentanyl and sufentanil. Microsomes prepared from different human liver samples were compared for their abilities to metabolize fentanyl, sufentanil and alfentanil, and it was found that disappearance of the three substrates was well correlated with immunoreactive CYP3A4 contents but not with other CYPs, including CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2D6 and CYP2E1. Specific known inhibitors of CYP enzymes gave similar results, whereas the use of recombinant human CYP enzymes expressed in yeast provided information about the possible involvement of other CYPs than CYP3A4 in the biotransformation of fentanyl and sufentanil. The possible in vivo interaction of fentanyl and sufentanil with other drugs catalyzed by CYP3A4 is also discussed.


Subject(s)
Alfentanil/metabolism , Cytochrome P-450 Enzyme System/metabolism , Fentanyl/metabolism , Isoenzymes/metabolism , Microsomes, Liver/metabolism , Sufentanil/metabolism , Benzoflavones/pharmacology , Coumarins/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Ditiocarb/pharmacology , Humans , Immunoblotting , Isoenzymes/antagonists & inhibitors , Microsomes, Liver/enzymology , Saccharomyces cerevisiae/enzymology , Transfection
19.
Biol Neonate ; 72(3): 133-41, 1997.
Article in English | MEDLINE | ID: mdl-9303211

ABSTRACT

The placental transfer of three opioids used in peridural analgesia, fentanyl, alfentanil and sufentanil, and two reference substances, antipyrine and *H2O, was determined ex vivo in the human placental cotyledon system. (1) In the first set of experiments, the infusion rates were constant and fixed at physiological flow rates. Under these conditions, the magnitude of the materno-fetal transfer was in the following order: *H2O = antipyrine = fentanyl > alfentanil > sufentanil. No particular influence of molecular weight, lipophilia, pKa or the degree of ionization could be discerned. (2) In the second set of experiments, the influence of different flow rates, reflecting various pathophysiological conditions, was examined. There was a linear relationship between the maternal flow and the materno-fetal transfer of the three opioids. On the other hand, for antipyrine and tritiated water, the relationship was logarithmic, a difference attributed to the marked lipophilia of the opioids. (3) At high maternal flow rates, saturation was observed for all five substances due to the short duration of contact with the membrane. There were logarithmic relationships between the maternal flow and the materno-fetal transfer. (4) These findings emphasize the importance of the lipophilic and hydrophilic characteristics of drugs on placental transfer, especially in the event of fluctuations in maternal flow.


Subject(s)
Alfentanil/metabolism , Analgesics, Opioid/metabolism , Fentanyl/metabolism , Maternal-Fetal Exchange/physiology , Placenta/blood supply , Sufentanil/metabolism , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antipyrine/metabolism , Biological Transport , Blood Flow Velocity , Female , Humans , In Vitro Techniques , Perfusion , Pregnancy , Reference Standards
20.
J Pharmacol Exp Ther ; 278(3): 1121-7, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8819494

ABSTRACT

In C6 glioma cells stably expressing a homogeneous population of the cloned rat mu opioid receptor, the binding affinities of opioid agonists and subsequent activation of G protein were examined. Opioid receptor number in membranes of these cells was high (10-30 pmol/mg protein [3H]diprenorphine binding sites). Opioids were found to bind to the receptor with high affinity [Tyr-D-Ala-Gly-(Me)Phe-Gly-ol (DAMGO) 0.23 nM; sufentanil 0.034 nM; morphine 0.16 nM]. Activation of G protein by opioid agonists was examined by measuring the stimulation of guanosine-5'-O-(3-[35S]thio)triphosphate ([35S]GTP gamma S) binding. Sufentanil increased [35S]GTP gamma S binding by 326% with an EC50 value of 2.39 nM. Agonist stimulation of [35S]GTP gamma S binding was stereoselective, naltrexone-reversible, and pertussis toxin-sensitive. The "intrinsic activity" of opioids at the mu receptor was reflected by the magnitude of agonist-mediated activation of G protein. The rank order of the stimulation of [35S]GTP gamma S binding was etonitazene = sufentanil = DAMGO = PLO17 = fentanyl > morphine > profadol > meperidine > butorphanol = nalbuphine = pentazocine > cyclazocine = nalorphine > levallorphan > naltrexone. High affinity binding of ligands to the mu opioid receptor was reduced by the addition of sodium and guanosine diphosphate at concentrations used in the [35S]GTP gamma S binding assay. Ligand affinity was reduced in a manner correlating with "intrinsic activity". DAMGO, 1229-fold, nalbuphine 35-fold, naltrexone, 3-fold. The results presented show that the stable expression of the rat mu opioid receptor in C6 cells provides an effective tool to examine opioid receptor signal transduction mechanisms and evaluate the activity of novel opioids at the mu receptor.


Subject(s)
Receptors, Opioid, mu/agonists , Animals , Binding, Competitive , Cell Membrane/metabolism , Enzyme Activation , GTP-Binding Proteins/metabolism , Glioma , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Radioligand Assay , Rats , Receptors, Opioid, mu/physiology , Recombinant Proteins , Signal Transduction , Sufentanil/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...